Open access peer-reviewed chapter - ONLINE FIRST

A Severe Clinical Example of Hypoxia; Sickle Cell Anemia

Written By

Büşra Tuğçe Akman

Submitted: 30 March 2023 Reviewed: 07 August 2023 Published: 27 September 2023

DOI: 10.5772/intechopen.1002900

Hypoxia - Recent Advances in the Field of Hypoxic and Ischemic Tissue Damage IntechOpen
Hypoxia - Recent Advances in the Field of Hypoxic and Ischemic Ti... Edited by Russell Peterson

From the Edited Volume

Hypoxia - Recent Advances in the Field of Hypoxic and Ischemic Tissue Damage [Working Title]

Dr. Russell Peterson and Dr. Russell Peterson

Chapter metrics overview

26 Chapter Downloads

View Full Metrics

Abstract

Sickle cell anemia is a disease in which the erythrocyte changes shape due to a mutation in the beta chain of hemoglobin causing vascular occlusion (vaso-occlusion) and clinical symptoms. In sickle cell patients, intermittent vascular occlusion leads to reperfusion injury associated with granulocyte accumulation and increased production of reactive oxygen species. Sickle cells adhere to endothelial cells and other blood cells more than normal erythrocytes in the microvascular circulation. The increase in thrombin and fibrin decreases the procoagulant activity of tissue factor, which triggers hypercoagulation. Where NO accumulates, oxidative stress reactions occur with vaso-occlusion. This results in decreased NO bioavailability and increased vascular dysfunction. Tissue damage due to vaso-occlusion causes the release of inflammatory mediators that trigger pain. Cytokines are released into the circulation by platelets, white blood cells, and endothelial cells. Patients with this condition are taken to the hospital with various syndromes such as occlusive crisis, acute chest syndrome, infection, multiple organ failure, and acute stroke. Sickle cell anemia effectively illustrates the severity of clinical manifestations caused by hypoxia.

Keywords

  • hemolysis
  • hypoxia
  • oxidative stress
  • sickle cell disease
  • vaso-occlusion

1. Introduction

Sickle cell disease (SCD) is the most common monogenic disorder [1]. With the “malaria hypothesis” formulated by Haldane in 1949 and Allison in 1954, the sickle cell trait is thought to protect against severe malaria [2]. The prevalence of the disease is high in large areas of sub-Saharan Africa, the Mediterranean basin, the Middle East, and India due to its natural protection against malaria [3]. In our country, Turkey, which is a Mediterranean country, the total number of patients with SCD is around 1200, and the frequency of the mutant Hb (HbS) of SCD is 0.03% [4]. Because of population movements, the distribution of sickle cell disease is widespread. According to estimates in the United States, about 100,000 people are thought to have the disease [5]. Every year, it is predicted that 300,000 babies are born with sickle cell anemia [6]. The vast majority of these births occur in three countries: Nigeria, the Democratic Republic of the Congo, and India. Sickle cell disease rates are expected to increase in the coming years. Over the last 40 years, survival rates have increased significantly in high-income countries with early screening, immunization, and use of antibiotics and hydroxyurea. In these countries, childhood deaths with sickle cell anemia are now close to deaths in the general population with a median survival of more than 60 years [7]. In Africa, where newborn screening and routine childhood vaccinations are absent and malaria, malnutrition, and poverty remain major problems, the mortality rate among children under 5 years of age with SCD can be up to 90% [8].

SCD occurs as a result of a point mutation in the β-globin chain of hemoglobin (replacement of glutamic acid at position 6 by valine). This mutant hemoglobin is called HbS. Conversion of hydrophilic glutamate to hydrophobic valine leads to polymerization of Hb and sickle erythrocytes (RBC) when deoxygenated [9, 10, 11]. While normal erythrocyte lifespan is 120 days, the lifespan of these deformed and hard sickled erythrocytes is shortened to 10–20 days. Because of their lack of flexibility, these erythrocytes cannot pass through microcapillaries resulting in extravascular and intravascular hemolysis, vaso-occlusion, abnormal erythrocyte-endothelial interactions, and ischemia-reperfusion injury. Persistent vaso-occlusion triggers further acute tissue hypoxia and chronic organ damage resulting in cycling that contributes to the progression of SCD. Hypoxia is a critical element in this cycle. Various physiological and pathological processes such as angiogenesis and inflammation have been shown to be regulated by hypoxia through a family of transcription factors known as hypoxia-inducible factors (HIFs), specifically HIF-la [12, 13]. Their effects include vaso-occlusion, increased production of free hemoglobin by lysis of erythrocytes, increased inflammation, platelet activation, increased erythrocyte adhesion to the vascular endothelium, and abnormal nitric oxide metabolism. Tissue damage increases with the interaction of sequestered neutrophils with the endothelium. These abnormalities lead to multisystem dysfunction with chronic inflammation, vascular damage, and anemia [14].

Advertisement

2. Pathophysiology of sickle cell disease

Hemoglobin consists of 2 alpha and 2 beta chains. HbS is the result of a specific point mutation in the HBB gene that encodes hemoglobin beta chains. It occurs with the replacement of glutamic acid in the seventh amino acid with valine. SCD is inherited autosomal recessively. It occurs when HbS is in a homozygous state (HbSS) or with some other HBB variants (Hb C, D or E) or when compound heterozygosity is present with beta thalassemia. The sections below describe the changes in the pathway from HbS development to SCD formation.

2.1 HbS polymerization and change of erythrocyte morphology

HbS polymerization is the initiating event from which all other pathophysiological changes occur [15]. Normal HbS is soluble in the cytoplasm of the RBC. Oxygen-free HbS polymerizes and precipitates inside the cell as a solid crystal. The sickle fibers that make up the polymerization of HbS initiate a series of events including damage to the RBC membrane, cellular dehydration, sickle-shaped cell transformation, and hemolysis. The formation of hemoglobin polymerization is an unstable event. Conditions such as hypoxia, 2,3-diphosphoglycerate (2,3-DPG) concentration, pH, temperature, and dehydration enhance the formation of intracellular polymerization [16, 17]. There are three important pathways that affect the dehydration of erythrocytes. These are KCl transport, the Gardos channel, and the Na-K pump system. It is reported in studies that the most important of these pathways is the Gardos channel. Even a minimal Gardos channel activation can lead to a potent and almost irreversible dehydration effect that promotes the sickling process [18]. The HbS polymer may cause reversible alterations to the RBC membrane. After repeated cycles, however, the red blood cell membrane becomes irreversibly damaged and sickle-shaped [19]. Sickle cells seen in peripheral smear are changed irreversibly [20]. Mature RBCs and reticulocytes can accumulate polymerized hemoglobin and transform into sickle cells. Immature, nucleated RBC progenitor cells do not sickle [21]. In sickle cells, abnormal globin chains cluster along the cell membrane and cause changes in cytoskeletal proteins such as band-3, ankyrin, and spectrin. Band-3 is an important integral membrane protein of erythrocytes and is required for the organization of the cytoskeleton, anion transport across the membrane, regulation of erythrocyte volume, and CO2 transport. Band-3 damage in SCD contributes to the sickling processes. Aggregation of globin chain clusters along the cytoskeletal membrane promotes IgG aggregation and triggers monocyte chemotaxis. This leads to the removal of aged erythrocytes from the circulation [22, 23].

2.2 Adhesion of sickle cells

The expression of typically underexpressed proteins such as phosphatidylserine (PS), basal cell adhesion molecule (B-CAM), integrin-associated protein (IAP), and intercellular adhesion molecule-4 (ICAM-4) increases when sickle erythrocytes’ membrane structure is damaged [24, 25, 26]. This situation facilitates the adhesion of erythrocytes to both endothelium and other blood cells and contributes to the pathogenesis of vaso-occlusive crises. According to research, the expression of adhesion molecules such as α4β1 (VLA-4) and CD36 is increased in the membrane of reticulocytes in SCD patients. This may play a role in the pathogenesis of cell adhesion and vascular occlusion [26]. As a result of the adhesion of the cells to the vascular endothelium, the transcription factor NF-KB is activated; oxygen radicals are released; and gene regulation and endothelial expression of adhesion molecules such as activated NF-κB, E-selectin, VCM-1, and ICAM-1 are increased [27].

2.3 Hemolysis and oxidative stress

In sickle cell disease, erythrocytes are exposed to intravascular and extravascular hemolysis and hemoglobin levels are 6–11 g/dl. While the rate of erythrocyte hemolysis is constant in patients with SCD during the stationary period, it increases during crisis. This increase varies according to the patient’s genotype (HbSS, HbSB, HbSC) and HbF level. In patients with high erythrocyte hemolysis and low Hb levels in the normal period, organ dysfunction in the form of vascular damage, endothelial dysfunction, and pulmonary hypertension associated with high pulse pressure, diastolic left heart disease, and renal dysfunction may develop in the future. In SCD patients, intravascular hemolysis releases free hemoglobin and heme; the plasma proteins haptoglobulin and hemopexin bind with these compounds, respectively, and remove them from the circulation. These protective mechanisms become dysfunctional in many pathological conditions, however, and hemopexin becomes completely depleted. Due to the absence of hemopexin, extremely high quantities of free heme accumulate in the blood. As a result, reactive oxygen molecules (ROS) are formed, which cause oxidative stress and cell damage [28]. In addition, free heme is lipophilic and can join with the lipid membranes of the vascular endothelium. As a result of the adhesion of activated endothelial cells with leukocytes, an inflammatory response is initiated that damages the endothelium and increases vascular permeability. Oxy-Hb released during hemolysis reacts with nitric oxide (NO) and lowers the plasma NO level. This condition causes endothelial dysfunction and has been associated with end-organ vasculopathy including leg ulcers, nephropathy, priapism, pulmonary hypertension, and death [28, 29, 30]. Erythrocytes release free hemoglobin and ADP, which stimulate platelet activation and contribute to vascular thrombosis and pulmonary hypertension by activating coagulation pathways [31, 32]. They also activate TLR4 and inflammasome signaling pathways as well as innate immune pathways [33]. RBC life is the greatest indicator for hemolysis and is substantially correlated with reticulocyte percentage and HbF level. Hb and hematocrit are not associated with RBC survival, however [34].

2.4 Inflammation and hypoxia

Erythrocyte injury-related molecules (eDAMP) released by hemolysis contribute to ischemia-reperfusion injury by supporting the formation and progression of inflammation. Heme and its oxidized form, hemin, are potent TLR4 agonists that contribute to the proinflammatory and procoagulant state in SCD patients. It has been reported that this condition is associated with activation of leukocytes, platelets, and endothelial cells, tissue factor release, cytokine storm, NO depletion, and ROS formation [35, 36, 37]. In addition, heme has also been shown to increase ADP- and epinephrine-dependent platelet aggregation [38]. Recent studies have shown that heme affects the migration and phagocytosis functions of monocytes, macrophages, and neutrophils leading to impaired bacterial clearance. Also, the G-protein-coupled receptor (GPBR) signaling pathway causes neutrophil migration, neutrophil extracellular trap (NET) production, oxidative damage, IL-8 production, and increased neutrophil survival [39, 40]. NETs are released from neutrophils under various inflammatory conditions and induce tissue damage by promoting the activation of innate immune responses. Repeated atherosclerosis and reperfusion attacks cause ischemia-reperfusion injury by promoting transient hypoxia, ROS generation, microvascular dysfunction, activation of innate and adaptive immune responses, and cell death [41, 42, 43]. This results in the activation of cell death programs such as ROS-induced damage to cellular proteins, lipids, DNA, and ribonucleic acids, apoptosis, necrosis, autophagy, and NETosis (the release of NETs by neutrophils). This in turn causes the release of various tissue and cell-derived DAMPs [43]. HIF1a, released after hypoxia in tissues, mediates adenosine release. Adenosine plays a central role in regulating the physiological response to hypoxia. It has long been known to mediate vasodilation, reduce inflammation, and mediate cell protection during ischemia-reperfusion injury in many organs. It was also revealed that adenosine stimulates RBCs to produce more 2,3-BPG, resulting in increased oxygen release from Hb. Although this contributes to the reduction of hypoxia in healthy individuals, it worsens the situation in SCD patients. This effect of adenosine is mediated by the G-protein-coupled adenosine receptor ADORA2B on the erythrocyte. The effect of adenosine on the ADORA2A receptor on invariant natural killer T (iNKT) cells has been shown to reduce pulmonary inflammation [44].

Advertisement

3. Clinical findings and management of sickle cell disease

The clinical manifestations of SCD may differ between major genotypes and even between patients with the same genotype. As a general rule, individuals with both SCD (homozygous HbS) and sickle-beta0 thalassemia have more severe symptoms than those with HbSC disease or sickle-beta+thalassemia, alone [1]. An exception is retinopathy, the most common complication in individuals with HbSC disease. Climate, meteorological changes, air quality, and infections are other environmental factors that affect the severity of the disease [45]. Acute and chronic complications are seen in sickle cell anemia. Painful crisis is the most common acute complication. Hemolytic crisis, splenic and hepatic sequestration crisis, acute chest syndrome, stroke, and priapism are other acute complications. Chronic complications are usually described in adults. Pulmonary hypertension, hepatopathy, kidney disease, splenomegaly, bone and joint deformities, retinopathy, iron deposition, and leg ulcers are its chronic complications. In addition, SCD patients have an increased susceptibility to infection. The life expectancy of patients with SCD has been shortened by at least 20 years, and the quality of life has decreased [14, 46]. Common complications and their management will be discussed below.

3.1 Acute and chronic pain

The most typical complication of SCD is severe intermittent acute pain (vaso-occlusive crisis), which is responsible for more than 70% of acute hospital visits in SCD patients [47]. Chronic daily pain increases with advanced age and occurs in 30–40% of adolescents and adults with SCD [48, 49]. Acute pain is largely associated with ischemia-reperfusion injury, tissue infarction, and vaso-occlusion of sickle red blood cells and occurs in an isolated anatomical site (e.g., arm, leg, back) or in multiple locations. Chronic pain may result from sensitization of the central and/or peripheral nervous system and is often accompanied by features of neuropathic pain [50, 51]. Disease complications such as avascular necrosis (hip, shoulder) and leg ulcers also cause chronic pain [52]. The aim of the intervention in acute pain is to relieve the patient’s pain or reduce it to an acceptable level to restore daily functions. Oral or intravenous (IV) opioids and IV hydration are first-line therapies for acute pain. Analgesics should be initiated within 30–60 minutes of triage [53]. Ketamine, a non-opioid analgesic, can be given as adjuvant therapy in acute pain resistant to opioids. Non-steroidal anti-inflammatory drugs (NSAIDs) are routinely used as adjuvant therapy for the management of acute pain [53, 54]. Red cell transfusion is indicated to rapidly reduce HbS levels or correct acute anemia (if splenic sequestration, hemolytic, or aplastic crisis are present). In patients with chronic pain, opioids can be given after the cost-benefit ratio has been calculated. In long-term use at increasing doses, the possibility of side effects including myocardial infarction, bone fracture, increased risk of motor vehicle impact, sexual dysfunction, and mortality should be considered. Trifluoperazine, an antipsychotic drug, serotonin, and norepinephrine reuptake inhibitors (SNRIs), gabapentinoids, and tricyclic antidepressants (TCAs) can be used for chronic pain [53, 55]. Acupuncture can also be tried [56].

3.2 Cardiopulmonary disease

Cardiopulmonary disease is associated with increased morbidity and mortality in individuals with SCD. Pulmonary arterial hypertension (PAH) is present in up to 10% of adults with SCD [57]. Pulmonary hypertension is defined as the measurement of mean pulmonary artery pressure (PAP) ≥25 mmHg at rest with right heart catheterization. In addition, the tricuspid regurgitation velocity (TRV) is measured as >2.5 m/s (s) by transthoracic echocardiography. Chronic intravascular hemolysis is the greatest risk factor for the development of PAH and leads to pulmonary arteriolar vasoconstriction and smooth muscle proliferation. Obstructive lung disease, restrictive lung disease, obstructive sleep apnea, or nocturnal hypoxemia are pulmonary disorders that may be present [58, 59, 60, 61]. Patients with SCD who have symptoms of cardiovascular illness, such as increased dyspnea, hypoxemia, or impaired exercise tolerance, should have a diagnostic ECHO and SFT. If there is snoring, witnessed apnea, breathing pauses, or hypoxia during sleep, daytime tiredness, or nocturnal enuresis, a sleep study should be performed in a diagnostic setting. Hydroxyurea or monthly red blood cell transfusions should be considered in the treatment of pulmonary hypertension. Endothelial receptor antagonists and prostanoids can be used in the selected patients. About 28% of children with SCD have asthma, associated with acute chest syndrome and higher mortality, as well as increased episodes of pain that may result from impaired oxygenation leading to sickling and vaso-occlusion [62, 63, 64]. First-line treatments include standard beta-adrenergic bronchodilators and oxygen as needed. When corticosteroids are indicated, they should be tapered off over a few days taking into account the risk of rebound SCD pain that may result from abrupt discontinuation of courses.

Patients with acute chest syndrome (AGS) usually present with a pneumonia-like clinical picture. It occurs due to vaso-occlusion in pulmonary vessels and manifests as hypoxia, fever, and new infiltration on chest X-ray. Since radiological findings may occur late, patients without infiltration should also be carefully evaluated with follow-up films. Although laboratory findings are not indicative, the hemoglobin concentration may drop suddenly. Although the most common etiological factor is infection, AGS can also occur from bone marrow embolism, intrapulmonary aggregates of communal cells, atelectasis, or pulmonary edema. It can very quickly lead to respiratory failure or death. It therefore requires urgent hospitalization. Treatment is antibiotics, oxygen support, and exchange transfusion in patients with respiratory failure. The risk of acute chest syndrome can be reduced with incentive spirometry and preoperative transfusions during hospitalization [14].

3.3 Central nervous system complications

Overt and silent cerebral infarctions cause significant morbidity in SCD patients. Eleven percent of patients by age 20 and 24% by age 45 will have had a detectable stroke [65]. Silent cerebral infarcts occur in 39% of adults at 18 years and > 50% at 30 years [66, 67]. Patients with both types of stroke have an increased risk of recurrent stroke [68]. Overt strokes occur in the great arteries, while silent cerebral infarcts occur in penetrating arteries. Overt strokes present as paresis, dysarthria or aphasia, seizures, sensory deficits, headaches, or altered consciousness. Silent infarctions are associated with cognitive deficits including low IQ and impaired academic performance. The diagnosis of overt stroke is made by evidence of acute infarction on brain MRI diffusion-weighted imaging and focal deficits on neurological examination. A silent cerebral infarction is defined by “fluid-attenuated inversion healing (FLAIR) MRI signal abnormality, at least 3 mm in one dimension and visible in 2 planes on T2-weighted images” and no defects on neurological examination [69]. Because silent cerebral infarcts are not clinically detectable, baseline brain MRI scanning is recommended in school-aged children with SCD [70]. Recent SCD clinical practice guidelines also recommend scanning the brain MRI in adults with SCD to facilitate rehabilitation services, patient, and family understanding of cognitive deficits, and further needs assessment [70]. Red blood cell exchange transfusion is the recommended treatment to rapidly reduce HbS below 30% in acute stroke [71]. A simple transfusion is usually given as an emergency measure when preparing for an exchange transfusion, however [70].

3.4 Renal disease

In SCD patients, glomerulopathy results from intravascular hemolysis and endothelial dysfunction in the renal cortex. Medullary hypoperfusion and ischemia also contribute to kidney disease in SCD causing hematuria, urinary concentration defects, and distal tubular dysfunction [72].

Glomerulopathy, characterized by hyperfiltration leading to albuminuria, is an early asymptomatic manifestation of SCD nephropathy. Hyperfiltration, defined as an absolute increase in the glomerular filtration rate, can be seen in 43% of children with SCD [73]. Albuminuria, defined by the presence of ≥30 mg/g urine albumin over 24 hours, was observed in 32% of adults with SCD [74]. About 20–40% of adults with SCD develop chronic kidney disease (CKD) and are at risk of developing end-stage renal disease (ESRD) [74]. Since microalbuminuria/proteinuria precedes CKD in SCD, annual urine microalbumin/protein screening is recommended starting at age 10 [46]. Kidney failure may be associated with the use of ketorolac in children with SCD hospitalized for pain. Although controlled clinical studies are lacking, hydroxyurea may be associated with improvements in glomerular hyperfiltration and urinary concentration ability in children with SCD [74, 75]. ACE-I or ARB therapy reduces microalbuminuria in patients with SCD [76]. ACE-I or ARB therapy has not been shown to improve kidney function or prevent CKD, however. Because of recent advances in renal graft survival and post-transplant mortality, renal transplantation should be considered in individuals with SCD and ESRD [77].

Advertisement

4. Modifying drugs used in treatment

Hydroxyurea, which is frequently used in SCD patients, is an antimetabolite that disrupts the DNA replication process. Stress induces fetal hemoglobin production through erythropoiesis, reduces inflammation, increases nitric oxide, and decreases cell adhesion. Other SCD-modifying drugs (i.e., L-glutamine, crizanlizumab, and voxelotor) were recently approved by the FDA. L-glutamine is required for the synthesis of nicotinamide adenine dinucleotide, glutathione, and arginine. This amino acid protects red blood cells against oxidative damage which is the basis for its proposed use in SCD. Crizanlizumab is a humanized monoclonal antibody that binds P-selectin and blocks the interaction of the adhesion molecule with its ligand (i.e., P-selectin glycoprotein ligand 1). P-selectin expression triggered by inflammation promotes adhesion of neutrophils, activated platelets, and sickle red blood cells to the endothelial surface and to each other thus promoting vaso-occlusion in SCD. Polymerization of the voxelotor Hb S in the deoxygenated state represents the first step in red blood cell sickling, which leads to reduced disfigurement of red blood cells and increased hemolysis. Voxelotor is the first allosteric modifier of HbS to increase oxygen affinity [78, 79, 80]. In addition to the modified drugs used in treatment, hematopoietic stem cell transplantation from HLA-matched sibling donors remains superior as the only curative treatment [81].

Advertisement

5. Conclusion

SCD, as a severe clinical example of hypoxia, progresses with complications including organ damage such as cardiopulmonary, cerebrovascular, and kidney disease resulting in increased morbidity and mortality. The survival of individuals with SCD is reduced compared to those without SCD, but the prognosis of SCD is steadily improving with neonatal screening, immunizations, antibiotics, hydroxyurea, and faster prevention and treatment. Where comprehensive care is available, the disease has evolved from a fatal pediatric illness to a chronic illness often associated with progressive deterioration in quality of life and organ function. Modifying drugs such as hydroxyurea L-glutamine, crizanlizumab, and voxelotor are frequently used for treatment. Fetal hemoglobin induction, sickle hemoglobin research on inhibition of polymerization, reduction of cell adhesion, and reduction of oxidative stress are continuing. Today, hematopoietic stem cell transplantation from HLA-matched sibling donors remains the only curative treatment.

References

  1. 1. Rees DC, Williams TN, Gladwin MT. Sickle-cell disease. Lancet. 2010;376:2018-2031
  2. 2. Elguero E, Délicat-Loembet LM, Rougeron V, et al. Malaria continues to select for sickle cell trait in Central Africa. Proceedings of the National Academy of Sciences of the United States of America. 2015;112:7051-7054
  3. 3. Piel FB, Patil AP, Howes RE, et al. Global distribution of the sickle cell gene and geographical confirmation of the malaria hypothesis. Nature Communications. 2010;1:104
  4. 4. Arcasoy A. Dünya’da ve Türkiye’de talasemi ve hemoglobinopatiler. In: Hemoglobinopati ve Talasemi Önlem Tanı Tedavi. Ulusal hemoglobinopati Konseyi Sağlık Bakanlığı. Baskı, Antalya; 2003. pp. 11-19
  5. 5. Hassell KL. Population estimates of sickle cell disease in the U.S. American Journal of Preventive Medicine. 2010;38(Suppl):S512-S521
  6. 6. Piel FB, Patil AP, Howes RE, et al. Global epidemiology of sickle haemoglobin in neonates: A contemporary geostatistical model-based map and population estimates. Lancet. 2013;381:142-151
  7. 7. Gardner K, Douiri A, Drasar E, et al. Survival in adults with sickle cell disease in a high-income setting. Blood. 2016;128:1436-1438
  8. 8. Grosse SD, Odame I, Atrash HK, Amendah DD, Piel FB, Williams TN. Sickle cell disease in Africa: A neglected cause of early childhood mortality. American Journal of Preventive Medicine. 2011;41(Suppl 4):S398-S405
  9. 9. Ingram VM. A specific chemical difference between the globins of normal human and sickle-cell anaemia haemoglobin. Nature. 1956;178:792-794
  10. 10. Chang JC, Kan YW. Antenatal diagnosis of sickle cell anaemia by direct analysis of the sickle mutation. Lancet. 1981;2:1127-1129
  11. 11. Hebbel RP. Beyond hemoglobin polymerization: The red blood cell membrane and sickle disease pathophysiology. Blood. 1991;77:214-237
  12. 12. Mucaj V, Shay JE, Simon MC. Effects of hypoxia and HIFs on cancer metabolism. International Journal of Hematology. 2012;95:464-470
  13. 13. Eltzschig HK, Carmeliet P. Hypoxia and inflammation. The New England Journal of Medicine. 2011;364:656-665
  14. 14. Kanter J, Kruse-Jarres R. Management of sickle cell disease from childhood through adulthood. Blood Reviews. 2013;27(6):279-287
  15. 15. Sundd P, Gladwin MT, Novelli EM. Pathophysiology of sickle cell disease. Annual Review of Pathology. 2019;14:263
  16. 16. Harmening DM. Hemoglobinopathies. Clinical Hematology and Fundamentals of Hemostasis. 3rd ed. Philadelphia. USA: FA Davis; 1997. pp. 173-192
  17. 17. Brittenham GM, Schechter AN, Noguchi CT. Hemoglobin S polymerization: Primary determinant of the hemolytic and clinical severity of the sickling syndromes. Blood. 1985;65:183-189
  18. 18. Brugnara c. Sickle cell dehydration: Pathophysiology and therapeutic applications. Clinical Hemorheology and Microcirculation. 2018;68(2-3):187-204
  19. 19. Serjeant GR, Petch MC, Serjeant BE. The in vivo sickle phenomenon: A reappraisal. The Journal of Laboratory and Clinical Medicine. 1973;81:850
  20. 20. Ballas SK, Eckman JR. Chapter 20: Sickle cell pain: biology, etiology, and treatment. In: Steinberg MH, Forget BG, Higgs DR, Weatherall DJ, editors. Disorders of Hemoglobin. 2nd ed. Cambridge: Cambridge University Press; 2009. p. 564
  21. 21. Grasso JA, Sullivan AL, Sullivan LW. Ultrastructural studies of the bone marrow in sickle cell anaemia. II. The morphology of erythropoietic cells and their response to deoxygenation in vitro. British Journal of Haematology. 1975;31:381
  22. 22. Platt OS, Falcone JF. Membrane protein interactions in sickle red blood cells: Evidence of abnormal protein 3 function. Blood. 1995;86(5):1992-1998
  23. 23. Bosman GJ. Erythrocyte aging in sickle cell disease. Cellular Molecular Biology (Noisy-le-grand). 2004;50(1):81-86
  24. 24. Bunn HF. Pathogenesis and treatment of sickle cell disease. The New England Journal of Medicine. 1997;337:762-769
  25. 25. Barabino GA, Platt MO, Kaul DK. Sickle cell biomechanics. Annual Review of Biomedical Engineering. 2010;12:345-367
  26. 26. Smith RA, Mankelow TJ, Drizou D, et al. Large red cell-derived membrane particles are major contributors to hypercoagulability in sickle cell disease. Scientific Reports. 2021;11:11035
  27. 27. Kaul DK, Finnegan E, Barabino GA. Sickle red cell-endothelium interactions. Microcirculation. 2009;16:97-111
  28. 28. Rp R, Bell L, Hillmen P, Gladwin MT. The clinical sequelae of intravascular hemolysis and extracellular plasma hemoglobin: A novel mechanism of human disease. Journal of the American Medical Association. 2005;293:1653-1662
  29. 29. Reiter CD, Wang X, Tanus-Santos JE, Hogg N, Cannon RO, et al. Cell-free hemoglobin limits nitric oxide bioavailability in sicklecell disease. Nature. 2002;8(12):1383-1389
  30. 30. Hsu LL, Champion HC, Campbell-Lee SA, Bivalacqua TJ, Manci EA, et al. Hemolysis in sickle cell mice causes pulmonary hypertension due to global impairment in nitric oxide bioavailability. Blood. 2007;109:3088-3098
  31. 31. Villagra J, Shiva S, Hunter LA, Machado RF, Gladwin MT, Kato GJ. 2007 platelet activation in patients with sickle disease, hemolysis-associated pulmonary hypertension, and nitric oxide scavenging by cell-free hemoglobin. Blood. 2007;110:2166-2172
  32. 32. Kı A. Hypercoagulability and thrombotic complications in hemolytic anemias. Haematologica. 2009;94(11):1481-1484
  33. 33. Gladwin MT, Ofori-Acquah SF. Erythroid DAMps drive inflammation in ScD. Blood. 2014;123:3689-3690
  34. 34. Hebbel RP. Reconstructing sickle cell disease: A data-based analysis of the “hyperhemolysis paradigm” for pulmonary hypertension from the perspective of evidence-based medicine. American Journal of Hematology. 2011;86:123
  35. 35. Serjeant GR, Serjeant BE, Milner PF. The irreversibly sickled cell; a determinant of haemolysis in sickle cell anaemia. British Journal of Haematology. 1969;17:527
  36. 36. Allali S, Maciel TT, Hermine O, Montalembert M. Innate immune cells, major protagonists of sickle cell disease pathophysiology. Haematologica. 2020;105(2):273-283
  37. 37. Dutra FF, Bozza MT. Heme on innate immunity and inflammation. Frontiers in Pharmacology. 2014;5:115
  38. 38. Malik Z, Creter D, Cohen A, Djaldetti M. Haemin affects platelet aggregation and lymphocyte mitogenicity in whole blood incubations. Cytobios. 1983;38:33-38
  39. 39. Kono M, Saigo K, Takagi Y, Takahashi T, Kawauchi S, et al. Heme-related molecules induce rapid production of neutrophil extracellular traps. Transfusion. 2014;54:2811-2819
  40. 40. Graca-Souza A, Arruda MA, de Freitas MS, Barja-Fidalgo C, Oliveira PL. Neutrophil activation by heme: Implications for inflammatory processes. Blood. 2002;99:4160-4165
  41. 41. Belcher JD, Bryant CJ, Nguyen J, Bowlin PR, Kielbik MC, et al. Transgenic sickle mice have vascular inflammation. Blood. 2003;101:3953-3959
  42. 42. Kaul DK, Rp H. Hypoxia/reoxygenation causes inflammatory response in transgenic sickle mice but not in normal mice. Journal of Clinical Investment. 2000;106:411-420
  43. 43. Eltzschig HK, Eckle T. Ischemia and reperfusion-from mechanism to translation. Nature. 2011;17:1391-1401
  44. 44. Sun K, Xia Y. New insights into sickle cell disease: A disease of hypoxia. Current Opinion in Hematology. 2013;20(3):215-221
  45. 45. Piel FB, Steinberg MH, Rees DC. Sickle cell disease. The New England Journal of Medicine. 2017;376(16):1561-1573
  46. 46. Yawn BP, Buchanan GR, Afenyi-Annan AN, Ballas SK, Hassell KL, James AH, et al. Management of sickle cell disease: Summary of the 2014 evidence-based report by expert panel members. JAMA. 2014;312(10):1033-1048
  47. 47. Brousseau DC, Owens PL, Mosso AL, Panepinto JA, Steiner CA. Acute care utilization and rehospitalizations for sickle cell disease. Journal of the American Medical Association. 2010;303(13):1288-1294
  48. 48. Sil S, Cohen LL, Dampier C. Psychosocial and functional outcomes in youth with chronic sickle cell pain. The Clinical Journal of Pain. 2016;32(6):527-533
  49. 49. Smith WR, Penberthy LT, Bovbjerg VE, McClish DK, Roberts JD, Dahman B, et al. Daily assessment of pain in adults with sickle cell disease. Annals of Internal Medicine. 2008;148(2):94-101
  50. 50. Ballas SK, Darbari DS. Neuropathy, neuropathic pain, and sickle cell disease. American Journal of Hematology. 2013;88(11):927-929
  51. 51. Sharma D, Brandow AM. Neuropathic pain in individuals with sickle cell disease. Neuroscience Letters. 2020;714:134445
  52. 52. Dampier C, Palermo TM, Darbari DS, Hassell K, Smith W, Zempsky W. AAPT diagnostic criteria for chronic sickle cell disease pain. The Journal of Pain. 2017;18(5):490-498
  53. 53. Brandow AM, Carroll CP, Creary S, et al. American Society of Hematology 2020 guidelines for sickle cell disease: Management of acute and chronic pain. Blood Advances. 2020;4(12):2656-2701
  54. 54. Lubega FA, DeSilva MS, Munube D, Nkwine R, Tumukunde J, Agaba PK, et al. Low dose ketamine versus morphine for acute severe vaso occlusive pain in children: A randomized controlled trial. Scandinavian Journal of Pain. 2018;18(1):19-27
  55. 55. Molokie RE, Wilkie DJ, Wittert H, Suarez ML, Yao Y, Zhao Z, et al. Mechanism-driven phase I translational study of trifluoperazine in adults with sickle cell disease. European Journal of Pharmacology. 2014;723:419-424
  56. 56. Mahmood LA, Reece-Stremtan S, Idiokitas R, Martin B, Margulies S, Hardy SJ, et al. Acupuncture for pain management in children with sickle cell disease. Complementary Therapies in Medicine. 2020;49:102287
  57. 57. Mehari A, Alam S, Tian X, Cuttica MJ, Barnett CF, Miles G, et al. Hemodynamic predictors of mortality in adults with sickle cell disease. American Journal of Respiratory and Critical Care Medicine. 2013;187(8):840-847
  58. 58. Cohen RT, Strunk RC, Rodeghier M, Rosen CL, Kirkham FJ, Kirkby J, et al. Pattern of lung function is not associated with prior or future morbidity in children with sickle cell anemia. Annals of the American Thoracic Society. 2016;13(8):1314-1323
  59. 59. Kassim AA, Payne AB, Rodeghier M, Macklin EA, Strunk RC, DeBaun MR. Low forced expiratory volume is associated with earlier death in sickle cell anemia. Blood. 2015;126(13):1544-1550
  60. 60. Sharma S, Efird JT, Knupp C, Kadali R, Liles D, Shiue K, et al. Sleep disorders in adult sickle cell patients. Journal of Clinical Sleep Medicine. 2015;11(3):219-223
  61. 61. Rosen CL, Debaun MR, Strunk RC, Redline S, Seicean S, Craven DI, et al. Obstructive sleep apnea and sickle cell anemia. Pediatrics. 2014;134(2):273-281
  62. 62. Knight-Madden JM, Barton-Gooden A, Weaver SR, Reid M, Greenough A. Mortality, asthma, smoking and acute chest syndrome in young adults with sickle cell disease. Lung. 2013;191(1):95-100
  63. 63. Strunk RC, Cohen RT, Cooper BP, Rodeghier M, Kirkham FJ, Warner JO, et al. Wheezing symptoms and parental asthma are associated with a physician diagnosis of asthma in children with sickle cell anemia. The Journal of Pediatrics. 2014;164(4):821-771
  64. 64. Takahashi T, Okubo Y, Handa A. Acute chest syndrome among children hospitalized with vaso-occlusive crisis: A nationwide study in the United States. Pediatric Blood & Cancer. 2018;65(3):e26885
  65. 65. Ohene-Frempong K, Weiner SJ, Sleeper LA, Miller ST, Embury S, Moohr JW, et al. Cerebrovascular accidents in sickle cell disease: Rates and risk factors. Blood. 1998;91(1):288-294
  66. 66. Bernaudin F, Verlhac S, Arnaud C, Kamdem A, Vasile M, Kasbi F, et al. Chronic and acute anemia and extracranial internal carotid stenosis are risk factors for silent cerebral infarcts in sickle cell anemia. Blood. 2015;125(10):1653-1661
  67. 67. Kassim AA, Pruthi S, Day M, Rodeghier M, Gindville MC, Brodsky MA, et al. Silent cerebral infarcts and cerebral aneurysms are prevalent in adults with sickle cell anemia. Blood. 2016;127(16):2038-2040
  68. 68. Powars D, Wilson B, Imbus C, Pegelow C, Allen J. The natural history of stroke in sickle cell disease. The American Journal of Medicine. 1978;65(3):461-471
  69. 69. DeBaun MR, Gordon M, McKinstry RC, Noetzel MJ, White DA, Sarnaik SA, et al. Controlled trial of transfusions for silent cerebral infarcts in sickle cell anemia. The New England Journal of Medicine. 2014;371(8):699-710
  70. 70. DeBaun MR, Jordan LC, King AA, Schatz J, Vichinsky E, Fox CK, et al. American Society of Hematology 2020 guidelines for sickle cell disease: Prevention, diagnosis, and treatment of cerebrovascular disease in children and adults. Blood Advances. 2020;4(8):1554-1588
  71. 71. Hulbert ML, Scothorn DJ, Panepinto JA, Scott JP, Buchanan GR, Sarnaik S, et al. Exchange blood transfusion compared with simple transfusion for first overt stroke is associated with a lower risk of subsequent stroke: A retrospective cohort study of 137 children with sickle cell anemia. The Journal of Pediatrics. 2006;149(5):710-712
  72. 72. Cazenave M, Audard V, Bertocchio JP, Habibi A, Baron S, Prot-Bertoye C, et al. Tubular acidification defect in adults with sickle cell disease. Clinical Journal of the American Society of Nephrology. 2020;15(1):16-24
  73. 73. Lebensburger JD, Aban I, Pernell B, Kasztan M, Feig DI, Hilliard LM, et al. Hyperfiltration during early childhood precedes albuminuria in pediatric sickle cell nephropathy. American Journal of Hematology. 2019;94(4):417-423
  74. 74. Niss O, Lane A, Asnani MR, Yee ME, Raj A, Creary S, et al. Progression of albuminuria in patients with sickle cell anemia: A multicenter, longitudinal study. Blood Advances. 2020;4(7):1501-1511
  75. 75. Zahr RS, Hankins JS, Kang G, Li C, Wang WC, Lebensburger J, et al. Hydroxyurea prevents onset and progression of albuminuria in children with sickle cell anemia. American Journal of Hematology. 2019;94(1):E27-E29
  76. 76. Quinn CT, Saraf SL, Gordeuk VR, Fitzhugh CD, Creary SE, Bodas P, et al. Losartan for the nephropathy of sickle cell anemia: A phase-2, multicenter trial. American Journal of Hematology. 2017;92(9):E520-E528
  77. 77. Gérardin C, Moktefi A, Couchoud C, Duquesne A, Ouali N, Gataut P, et al. Survival and specific outcome of sickle cell disease patients after renal transplantation. British Journal of Haematology. 2019;187(5):676-680
  78. 78. Ataga KI, Kutlar A, Kanter J, Liles D, Cancado R, Friedrisch J, et al. Crizanlizumab for the prevention of pain crises in sickle cell disease. The New England Journal of Medicine. 2017;376(5):429-439
  79. 79. Niihara Y, Miller ST, Kanter J, Lanzkron S, Smith WR, Hsu LL, et al. A phase 3 trial of L-glutamine in sickle cell disease. The New England Journal of Medicine. 2018;379(3):226-235
  80. 80. Vichinsky E, Hoppe CC, Ataga KI, Ware RE, Nduba V, El-Beshlawy A, et al. A phase 3 randomized trial of voxelotor in sickle cell disease. The New England Journal of Medicine. 2019;381(6):509-519
  81. 81. Gluckman E, Cappelli B, Bernaudin F, Labopin M, Volt F, Carreras J, et al. Sickle cell disease: An international survey of results of HLA-identical sibling hematopoietic stem cell transplantation. Blood. 2017;129(11):1548-1556

Written By

Büşra Tuğçe Akman

Submitted: 30 March 2023 Reviewed: 07 August 2023 Published: 27 September 2023